1
|
Ofori‐Kwafo A, Sigdel I, Al Mamun E, Zubcevic J, Tang Y. Gut-on-a-chip platforms: Bridging in vitro and in vivo models for advanced gastrointestinal research. Physiol Rep 2025; 13:e70356. [PMID: 40323242 PMCID: PMC12051376 DOI: 10.14814/phy2.70356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/11/2025] [Accepted: 04/22/2025] [Indexed: 05/08/2025] Open
Abstract
The gastrointestinal (GI) tract plays a critical role in nutrient absorption, immune responses, and overall health. Traditional models such as two-dimensional cell cultures have provided valuable insights but fail to replicate the dynamic and complex microenvironment of the human gut. Gut-on-a-chip platforms, which incorporate cells located in the gut into microfluidic devices that simulate peristaltic motion and fluid flow, represent a significant advancement in modeling GI physiology and diseases. This review discusses the evolution of gut-on-a-chip technology, from simple cellular mono-cultures models to more sophisticated systems incorporating bi-cultures and tri-cultures that enable studies of drug metabolism, disease modeling, and gut-microbiome interactions. Although challenges remain, including maintaining long-term cell viability and replicating immune responses, these platforms hold great potential for advancing personalized medicine and improving drug discovery efforts targeting gastrointestinal disorders.
Collapse
Affiliation(s)
- Awurama Ofori‐Kwafo
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Indira Sigdel
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Earshed Al Mamun
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| | - Jasenka Zubcevic
- University of South Florida Center for Microbiome ResearchMicrobiomes InstituteTampaFloridaUSA
- Department of Neurosurgery and Brain RepairUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Yuan Tang
- Department of Bioengineering, College of EngineeringUniversity of ToledoToledoOhioUSA
| |
Collapse
|
2
|
Brandauer K, Schweinitzer S, Lorenz A, Krauß J, Schobesberger S, Frauenlob M, Ertl P. Advances of dual-organ and multi-organ systems for gut, lung, skin and liver models in absorption and metabolism studies. LAB ON A CHIP 2025; 25:1384-1403. [PMID: 39973270 DOI: 10.1039/d4lc01011f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Drug development is a costly and timely process with high risks of failure during clinical trials. Although in vitro tissue models have significantly advanced over the years, thus fostering a transition from animal-derived models towards human-derived models, failure rates still remain high. Current cell-based assays are still not able to provide an accurate prediction of the clinical success or failure of a drug candidate. To overcome the limitations of current methods, a variety of microfluidic systems have been developed as powerful tools that are capable of mimicking (micro)physiological conditions more closely by integrating physiological fluid flow conditions, mechanobiological cues and concentration gradients, to name only a few. One major advantage of these biochip-based tissue cultures, however, is their ability to seamlessly connect different organ models, thereby allowing the study of organ-crosstalk and metabolic byproduct effects. This is especially important when assessing absorption, distribution, metabolism, and excretion (ADME) processes of drug candidates, where an interplay between various organs is a prerequisite. In the current review, a number of in vitro models as well as microfluidic dual- and multi-organ systems are summarized with a focus on absorption (skin, lung, gut) and metabolism (liver). Additionally, the advantage of multi-organ chips in identifying a drug's on and off-target toxicity is discussed. Finally, the potential high-throughput implementation and modular chip design of multi-organ-on-a-chip systems within the pharmaceutical industry is highlighted, outlining the necessity of reducing handling complexity.
Collapse
Affiliation(s)
- Konstanze Brandauer
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Sophie Schweinitzer
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Alexandra Lorenz
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Judith Krauß
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | | | - Martin Frauenlob
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| | - Peter Ertl
- Faculty of Technical Chemistry, TU Wien, Getreidemarkt 9, 1060 Vienna, Austria.
| |
Collapse
|
3
|
Deal H, Byrnes EM, Pandit S, Sheridan A, Brown AC, Daniele M. Injury-on-a-chip for modelling microvascular trauma-induced coagulation. LAB ON A CHIP 2025; 25:440-453. [PMID: 39763291 PMCID: PMC11704661 DOI: 10.1039/d4lc00471j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
Blood coagulation is a highly regulated injury response that features polymerization of fibrin fibers to prevent the passage of blood from a damaged vascular endothelium. A growing body of research seeks to monitor coagulation in microfluidic systems but fails to capture coagulation as a response to disruption of the vascular endothelium. Here we present a device that allows compression injury of a defined segment of a microfluidic vascular endothelium and the assessment of coagulation at the injury site. This pressure injury-on-a-chip (PINCH) device allows visualization of coagulation as the accumulation of fluorescent fibrin at injury sites. Quantification of fluorescent fibrin levels upstream of and at injury sites confirm that pre-treating vascular endothelium with fluid shear stress helps capture coagulation as an injury response. We leverage the PINCH devices to demonstrate the limited coagulation response of type A hemophiliacs and evaluate the performance of hemostatic microparticles and fibrinolytic nanoparticles. Our findings and the straightforward fabrication of the PINCH devices make it a promising choice for additional screening of hemostatic therapeutics.
Collapse
Affiliation(s)
- Halston Deal
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA.
| | - Elizabeth M Byrnes
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
| | - Sanika Pandit
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA.
| | - Anastasia Sheridan
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA.
| | - Ashley C Brown
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA.
| | - Michael Daniele
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina, Chapel Hill, 1840 Entrepreneur Dr., Raleigh, NC, 27695 USA.
- Comparative Medicine Institute, North Carolina State University, 1060 William Moore Dr., Raleigh, NC 27606, USA.
- Department of Electrical & Computer Engineering, North Carolina State University, 890 Oval Dr., Raleigh, NC, 27695 USA
| |
Collapse
|
4
|
Lopez Quezada L, Mba Medie F, Luu RJ, Gaibler RB, Gabriel EP, Rubio LD, Mulhern TJ, Marr EE, Borenstein JT, Fisher CR, Gard AL. Predicting Clinical Outcomes of SARS-CoV-2 Drug Efficacy with a High-Throughput Human Airway Microphysiological System. Adv Biol (Weinh) 2024; 8:e2300511. [PMID: 39123296 DOI: 10.1002/adbi.202300511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/23/2023] [Indexed: 08/12/2024]
Abstract
The average cost to bring a new drug from its initial discovery to a patient's bedside is estimated to surpass $2 billion and requires over a decade of research and development. There is a need for new drug screening technologies that can parse drug candidates with increased likelihood of clinical utility early in development in order to increase the cost-effectiveness of this pipeline. For example, during the COVID-19 pandemic, resources were rapidly mobilized to identify effective therapeutic treatments but many lead antiviral compounds failed to demonstrate efficacy when progressed to human trials. To address the lack of predictive preclinical drug screening tools, PREDICT96-ALI, a high-throughput (n = 96) microphysiological system (MPS) that recapitulates primary human tracheobronchial tissue,is adapted for the evaluation of differential antiviral efficacy of native SARS-CoV-2 variants of concern. Here, PREDICT96-ALI resolves both the differential viral kinetics between variants and the efficacy of antiviral compounds over a range of drug doses. PREDICT96-ALI is able to distinguish clinically efficacious antiviral therapies like remdesivir and nirmatrelvir from promising lead compounds that do not show clinical efficacy. Importantly, results from this proof-of-concept study track with known clinical outcomes, demonstrate the feasibility of this technology as a prognostic drug discovery tool.
Collapse
Affiliation(s)
| | | | - Rebeccah J Luu
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| | | | | | - Logan D Rubio
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| | | | | | | | | | - Ashley L Gard
- Bioengineering Division, Draper, Cambridge, MA, 02139, USA
| |
Collapse
|
5
|
Padmanaban AM, Ganesan K, Ramkumar KM. A Co-Culture System for Studying Cellular Interactions in Vascular Disease. Bioengineering (Basel) 2024; 11:1090. [PMID: 39593750 PMCID: PMC11591305 DOI: 10.3390/bioengineering11111090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) are leading causes of morbidity and mortality globally, characterized by complications such as heart failure, atherosclerosis, and coronary artery disease. The vascular endothelium, forming the inner lining of blood vessels, plays a pivotal role in maintaining vascular homeostasis. The dysfunction of endothelial cells contributes significantly to the progression of CVDs, particularly through impaired cellular communication and paracrine signaling with other cell types, such as smooth muscle cells and macrophages. In recent years, co-culture systems have emerged as advanced in vitro models for investigating these interactions and mimicking the pathological environment of CVDs. This review provides an in-depth analysis of co-culture models that explore endothelial cell dysfunction and the role of cellular interactions in the development of vascular diseases. It summarizes recent advancements in multicellular co-culture models, their physiological and therapeutic relevance, and the insights they provide into the molecular mechanisms underlying CVDs. Additionally, we evaluate the advantages and limitations of these models, offering perspectives on how they can be utilized for the development of novel therapeutic strategies and drug testing in cardiovascular research.
Collapse
Affiliation(s)
- Abirami M. Padmanaban
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India;
| | - Kumar Ganesan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong 999077, China;
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India;
| |
Collapse
|
6
|
Kim J, Ro J, Cho YK. Vascularized platforms for investigating cell communication via extracellular vesicles. BIOMICROFLUIDICS 2024; 18:051504. [PMID: 39323481 PMCID: PMC11421861 DOI: 10.1063/5.0220840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
The vascular network plays an essential role in the maintenance of all organs in the body via the regulated delivery of oxygen and nutrients, as well as tissue communication via the transfer of various biological signaling molecules. It also serves as a route for drug administration and affects pharmacokinetics. Due to this importance, engineers have sought to create physiologically relevant and reproducible vascular systems in tissue, considering cell-cell and extracellular matrix interaction with structural and physical conditions in the microenvironment. Extracellular vesicles (EVs) have recently emerged as important carriers for transferring proteins and genetic material between cells and organs, as well as for drug delivery. Vascularized platforms can be an ideal system for studying interactions between blood vessels and EVs, which are crucial for understanding EV-mediated substance transfer in various biological situations. This review summarizes recent advances in vascularized platforms, standard and microfluidic-based techniques for EV isolation and characterization, and studies of EVs in vascularized platforms. It provides insights into EV-related (patho)physiological regulations and facilitates the development of EV-based therapeutics.
Collapse
|
7
|
Hall E, Mendiola K, Lightsey NK, Hanjaya-Putra D. Mimicking blood and lymphatic vasculatures using microfluidic systems. BIOMICROFLUIDICS 2024; 18:031502. [PMID: 38726373 PMCID: PMC11081709 DOI: 10.1063/5.0175154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/04/2024] [Indexed: 05/12/2024]
Abstract
The role of the circulatory system, containing the blood and lymphatic vasculatures, within the body, has become increasingly focused on by researchers as dysfunction of either of the systems has been linked to serious complications and disease. Currently, in vivo models are unable to provide the sufficient monitoring and level of manipulation needed to characterize the fluidic dynamics of the microcirculation in blood and lymphatic vessels; thus in vitro models have been pursued as an alternative model. Microfluidic devices have the required properties to provide a physiologically relevant circulatory system model for research as well as the experimental tools to conduct more advanced research analyses of microcirculation flow. In this review paper, the physiological behavior of fluid flow and electrical communication within the endothelial cells of the systems are detailed and discussed to highlight their complexities. Cell co-culturing methods and other relevant organ-on-a-chip devices will be evaluated to demonstrate the feasibility and relevance of the in vitro microfluidic model. Microfluidic systems will be determined as a noteworthy model that can display physiologically relevant flow of the cardiovascular and lymphatic systems, which will enable researchers to investigate the systems' prevalence in diseases and identify potential therapeutics.
Collapse
Affiliation(s)
- Eva Hall
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | | - N. Keilany Lightsey
- Aerospace and Mechanical Engineering, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | | |
Collapse
|
8
|
Żuchowska A, Baranowska P, Flont M, Brzózka Z, Jastrzębska E. Review: 3D cell models for organ-on-a-chip applications. Anal Chim Acta 2024; 1301:342413. [PMID: 38553129 DOI: 10.1016/j.aca.2024.342413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 04/02/2024]
Abstract
Two-dimensional (2D) cultures do not fully reflect the human organs' physiology and the real effectiveness of the used therapy. Therefore, three-dimensional (3D) models are increasingly used in bioanalytical science. Organ-on-a-chip systems are used to obtain cellular in vitro models, better reflecting the human body's in vivo characteristics and allowing us to obtain more reliable results than standard preclinical models. Such 3D models can be used to understand the behavior of tissues/organs in response to selected biophysical and biochemical factors, pathological conditions (the mechanisms of their formation), drug screening, or inter-organ interactions. This review characterizes 3D models obtained in microfluidic systems. These include spheroids/aggregates, hydrogel cultures, multilayers, organoids, or cultures on biomaterials. Next, the methods of formation of different 3D cultures in Organ-on-a-chip systems are presented, and examples of such Organ-on-a-chip systems are discussed. Finally, current applications of 3D cell-on-a-chip systems and future perspectives are covered.
Collapse
Affiliation(s)
- Agnieszka Żuchowska
- Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland
| | - Patrycja Baranowska
- Center for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poleczki 19, 02-822, Warsaw, Poland
| | - Magdalena Flont
- Center for Advanced Materials and Technologies CEZAMAT, Warsaw University of Technology, Poleczki 19, 02-822, Warsaw, Poland
| | - Zbigniew Brzózka
- Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland
| | - Elżbieta Jastrzębska
- Medical Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664, Warsaw, Poland.
| |
Collapse
|
9
|
Li H, Shang Y, Zeng J, Matsusaki M. Technology for the formation of engineered microvascular network models and their biomedical applications. NANO CONVERGENCE 2024; 11:10. [PMID: 38430377 PMCID: PMC10908775 DOI: 10.1186/s40580-024-00416-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/15/2024] [Indexed: 03/03/2024]
Abstract
Tissue engineering and regenerative medicine have made great progress in recent decades, as the fields of bioengineering, materials science, and stem cell biology have converged, allowing tissue engineers to replicate the structure and function of various levels of the vascular tree. Nonetheless, the lack of a fully functional vascular system to efficiently supply oxygen and nutrients has hindered the clinical application of bioengineered tissues for transplantation. To investigate vascular biology, drug transport, disease progression, and vascularization of engineered tissues for regenerative medicine, we have analyzed different approaches for designing microvascular networks to create models. This review discusses recent advances in the field of microvascular tissue engineering, explores potential future challenges, and offers methodological recommendations.
Collapse
Affiliation(s)
- He Li
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yucheng Shang
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Jinfeng Zeng
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
10
|
Mansouri M, Hughes AR, Audi LA, Carter AE, Vidas JA, McGrath JL, Abhyankar VV. Transforming Static Barrier Tissue Models into Dynamic Microphysiological Systems. J Vis Exp 2024:10.3791/66090. [PMID: 38436378 PMCID: PMC11096840 DOI: 10.3791/66090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2024] Open
Abstract
Microphysiological systems are miniaturized cell culture platforms used to mimic the structure and function of human tissues in a laboratory setting. However, these platforms have not gained widespread adoption in bioscience laboratories where open-well, membrane-based approaches serve as the gold standard for mimicking tissue barriers, despite lacking fluid flow capabilities. This issue can be primarily attributed to the incompatibility of existing microphysiological systems with standard protocols and tools developed for open-well systems. Here, we present a protocol for creating a reconfigurable membrane-based platform with an open-well structure, flow enhancement capability, and compatibility with conventional protocols. This system utilizes a magnetic assembly approach that enables reversible switching between open-well and microfluidic modes. With this approach, users have the flexibility to begin an experiment in the open-well format using standard protocols and add or remove flow capabilities as needed. To demonstrate the practical usage of this system and its compatibility with standard techniques, an endothelial cell monolayer was established in an open-well format. The system was reconfigured to introduce fluid flow and then switched to the open-well format to conduct immunostaining and RNA extraction. Due to its compatibility with conventional open-well protocols and flow enhancement capability, this reconfigurable design is expected to be adopted by both engineering and bioscience laboratories.
Collapse
Affiliation(s)
- Mehran Mansouri
- Department of Biomedical Engineering, Rochester Institute of Technology
| | - Aidan R Hughes
- Department of Biomedical Engineering, Rochester Institute of Technology
| | - Lauren A Audi
- Department of Biomedical Engineering, Rochester Institute of Technology
| | - Anna E Carter
- Department of Biomedical Engineering, Rochester Institute of Technology
| | - Justin A Vidas
- Department of Biomedical Engineering, Rochester Institute of Technology
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester
| | - Vinay V Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology;
| |
Collapse
|
11
|
Maurissen TL, Spielmann AJ, Schellenberg G, Bickle M, Vieira JR, Lai SY, Pavlou G, Fauser S, Westenskow PD, Kamm RD, Ragelle H. Modeling early pathophysiological phenotypes of diabetic retinopathy in a human inner blood-retinal barrier-on-a-chip. Nat Commun 2024; 15:1372. [PMID: 38355716 PMCID: PMC10866954 DOI: 10.1038/s41467-024-45456-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
Diabetic retinopathy (DR) is a microvascular disorder characterized by inner blood-retinal barrier (iBRB) breakdown and irreversible vision loss. While the symptoms of DR are known, disease mechanisms including basement membrane thickening, pericyte dropout and capillary damage remain poorly understood and interventions to repair diseased iBRB microvascular networks have not been developed. In addition, current approaches using animal models and in vitro systems lack translatability and predictivity to finding new target pathways. Here, we develop a diabetic iBRB-on-a-chip that produces pathophysiological phenotypes and disease pathways in vitro that are representative of clinical diagnoses. We show that diabetic stimulation of the iBRB-on-a-chip mirrors DR features, including pericyte loss, vascular regression, ghost vessels, and production of pro-inflammatory factors. We also report transcriptomic data from diabetic iBRB microvascular networks that may reveal drug targets, and examine pericyte-endothelial cell stabilizing strategies. In summary, our model recapitulates key features of disease, and may inform future therapies for DR.
Collapse
Affiliation(s)
- Thomas L Maurissen
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Alena J Spielmann
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Gabriella Schellenberg
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Marc Bickle
- Roche Pharma Research and Early Development, Institute of Human Biology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Jose Ricardo Vieira
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Si Ying Lai
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Georgios Pavlou
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sascha Fauser
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Peter D Westenskow
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Roger D Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Héloïse Ragelle
- Roche Pharma Research and Early Development, Cardiovascular, Metabolism, Immunology, Infectious Diseases and Ophthalmology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| |
Collapse
|
12
|
Soragni C, Queiroz K, Ng CP, Stok A, Olivier T, Tzagkaraki D, Heijmans J, Suijker J, de Ruiter SPM, Olczyk A, Bokkers M, Schavemaker F, Trietsch SJ, Lanz HL, Vulto P, Joore J. Phenotypic screening in Organ-on-a-Chip systems: a 1537 kinase inhibitor library screen on a 3D angiogenesis assay. Angiogenesis 2024; 27:37-49. [PMID: 37493987 PMCID: PMC10881651 DOI: 10.1007/s10456-023-09888-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/13/2023] [Indexed: 07/27/2023]
Abstract
Modern drug development increasingly requires comprehensive models that can be utilized in the earliest stages of compound and target discovery. Here we report a phenotypic screening exercise in a high-throughput Organ-on-a-Chip setup. We assessed the inhibitory effect of 1537 protein kinase inhibitors in an angiogenesis assay. Over 4000 micro-vessels were grown under perfusion flow in microfluidic chips, exposed to a cocktail of pro-angiogenic factors and subsequently exposed to the respective kinase inhibitors. Efficacy of compounds was evaluated by reduced angiogenic sprouting, whereas reduced integrity of the main micro-vessel was taken as a measure for toxicity. The screen yielded 53 hits with high anti-angiogenicity and low toxicity, of which 44 were previously unassociated with angiogenic pathways. This study demonstrates that Organ-on-a-Chip models can be screened in high numbers to identify novel compounds and targets. This will ultimately reduce bias in early-stage drug development and increases probability to identify first in class compounds and targets for today's intractable diseases.
Collapse
Affiliation(s)
- Camilla Soragni
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
- Department of Cardiology, Maastricht University, Maastricht, The Netherlands
| | - Karla Queiroz
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Chee Ping Ng
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Arthur Stok
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Thomas Olivier
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Dora Tzagkaraki
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Jeroen Heijmans
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Johnny Suijker
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | | | | | - Marleen Bokkers
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | | | | | | | - Paul Vulto
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands
| | - Jos Joore
- MIMETAS BV, De Limes 7, 2342 DH, Oegstgeest, The Netherlands.
| |
Collapse
|
13
|
Rubio LD, McFarland KA, O'Seaghdha M, Williams C. A high throughput microphysiological model of prosthetic valve endocarditis for investigating factors that influence bacterial adhesion under fluid shear stress. Biochem Biophys Res Commun 2023; 686:149155. [PMID: 37926046 DOI: 10.1016/j.bbrc.2023.149155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 10/25/2023] [Indexed: 11/07/2023]
Abstract
Prosthetic heart valves are associated with almost one quarter of cases of infective endocarditis, a rare but serious condition with a staggering 25 % mortality rate. Without the endothelium of native valves, the risk of infection is exacerbated for implanted devices exposed to blood. There are currently no physiologically relevant in vitro or animal models of prosthetic valve endocarditis (PVE). Of particular importance, Staphylococcus aureus, a common agent of PVE, has demonstrated enhanced binding to blood plasma proteins (e.g., fibrinogen) and exposed matrix under fluid shear stress (FSS). An in vitro platform that mimics the multiple physiological determinants for S. aureus adhesion to prosthetic valve materials would facilitate the discovery of new treatments to minimize PVE. To this end, we developed a first-of-its-kind microphysiological model of PVE to study the effects of several key variables (endothelial cell coverage, fibrinogen deposition, surface treatments, and FSS) on S. aureus adhesion to bioprosthetic material surfaces. Our model demonstrated that viable endothelial monolayers diminished the deposition of fibrinogen and that fibrinogen was required for the subsequent adhesion of S. aureus to the bioprosthetic surface model. Next, we examined factors that affected endothelial cell coverage, such as FSS and glutaraldehyde, a common chemical treatment for bioprosthetic materials. In particular, glutaraldehyde treatment obstructed endothelialization of otherwise biocompatible collagen-coated surfaces, further enabling fibrinogen and S. aureus deposition. In future work, this model could impact multiple research areas, such as screening candidate bioprosthetic valve materials and new surface treatments to prevent PVE and further understanding host-pathogen interactions.
Collapse
Affiliation(s)
- Logan D Rubio
- Bioengineering Division, The Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Kirsty A McFarland
- Bioengineering Division, The Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | | | - Corin Williams
- Bioengineering Division, The Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States.
| |
Collapse
|
14
|
Shakeri A, Wang Y, Zhao Y, Landau S, Perera K, Lee J, Radisic M. Engineering Organ-on-a-Chip Systems for Vascular Diseases. Arterioscler Thromb Vasc Biol 2023; 43:2241-2255. [PMID: 37823265 PMCID: PMC10842627 DOI: 10.1161/atvbaha.123.318233] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 09/27/2023] [Indexed: 10/13/2023]
Abstract
Vascular diseases, such as atherosclerosis and thrombosis, are major causes of morbidity and mortality worldwide. Traditional in vitro models for studying vascular diseases have limitations, as they do not fully recapitulate the complexity of the in vivo microenvironment. Organ-on-a-chip systems have emerged as a promising approach for modeling vascular diseases by incorporating multiple cell types, mechanical and biochemical cues, and fluid flow in a microscale platform. This review provides an overview of recent advancements in engineering organ-on-a-chip systems for modeling vascular diseases, including the use of microfluidic channels, ECM (extracellular matrix) scaffolds, and patient-specific cells. We also discuss the limitations and future perspectives of organ-on-a-chip for modeling vascular diseases.
Collapse
Affiliation(s)
- Amid Shakeri
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Ying Wang
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Yimu Zhao
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Shira Landau
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Kevin Perera
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Jonguk Lee
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- KITE - Toronto Rehabilitation Institute, University Health Network, Toronto, Canada
| | - Milica Radisic
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto; Ontario, M5S 3E5; Canada
| |
Collapse
|
15
|
Fisher CR, Mba Medie F, Luu RJ, Gaibler RB, Mulhern TJ, Miller CR, Zhang CJ, Rubio LD, Marr EE, Vijayakumar V, Gabriel EP, Lopez Quezada L, Zhang CH, Anderson KS, Jorgensen WL, Alladina JW, Medoff BD, Borenstein JT, Gard AL. A High-Throughput, High-Containment Human Primary Epithelial Airway Organ-on-Chip Platform for SARS-CoV-2 Therapeutic Screening. Cells 2023; 12:2639. [PMID: 37998374 PMCID: PMC10669988 DOI: 10.3390/cells12222639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/25/2023] Open
Abstract
COVID-19 emerged as a worldwide pandemic in early 2020, and while the rapid development of safe and efficacious vaccines stands as an extraordinary achievement, the identification of effective therapeutics has been less successful. This process has been limited in part by a lack of human-relevant preclinical models compatible with therapeutic screening on the native virus, which requires a high-containment environment. Here, we report SARS-CoV-2 infection and robust viral replication in PREDICT96-ALI, a high-throughput, human primary cell-based organ-on-chip platform. We evaluate unique infection kinetic profiles across lung tissue from three human donors by immunofluorescence, RT-qPCR, and plaque assays over a 6-day infection period. Enabled by the 96 devices/plate throughput of PREDICT96-ALI, we also investigate the efficacy of Remdesivir and MPro61 in a proof-of-concept antiviral study. Both compounds exhibit an antiviral effect against SARS-CoV-2 in the platform. This demonstration of SARS-CoV-2 infection and antiviral dosing in a high-throughput organ-on-chip platform presents a critical capability for disease modeling and therapeutic screening applications in a human physiology-relevant in vitro system.
Collapse
Affiliation(s)
- Christine R. Fisher
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Felix Mba Medie
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Rebeccah J. Luu
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Robert B. Gaibler
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Thomas J. Mulhern
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Caitlin R. Miller
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Chelsea J. Zhang
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Logan D. Rubio
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Elizabeth E. Marr
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Vidhya Vijayakumar
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Elizabeth P. Gabriel
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Landys Lopez Quezada
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Chun-Hui Zhang
- Department of Chemistry, Yale University, New Haven, CT 06520, USA (W.L.J.)
| | - Karen S. Anderson
- Department of Pharmacology, Yale University, New Haven, CT 06520, USA;
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA
| | | | - Jehan W. Alladina
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (J.W.A.); (B.D.M.)
| | - Benjamin D. Medoff
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; (J.W.A.); (B.D.M.)
| | - Jeffrey T. Borenstein
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| | - Ashley L. Gard
- Bioengineering Division, Draper, Cambridge, MA 02139, USA; (C.R.F.); (F.M.M.); (R.J.L.); (R.B.G.); (T.J.M.); (V.V.); (E.P.G.); (L.L.Q.); (J.T.B.)
| |
Collapse
|
16
|
Jordan R, Ford-Scheimer SL, Alarcon RM, Atala A, Borenstein JT, Brimacombe KR, Cherry S, Clevers H, Davis MI, Funnell SGP, Gehrke L, Griffith LG, Grossman AC, Hartung T, Ingber DE, Kleinstreuer NC, Kuo CJ, Lee EM, Mummery CL, Pickett TE, Ramani S, Rosado-Olivieri EA, Struble EB, Wan Z, Williams MS, Hall MD, Ferrer M, Markossian S. Report of the Assay Guidance Workshop on 3-Dimensional Tissue Models for Antiviral Drug Development. J Infect Dis 2023; 228:S337-S354. [PMID: 37669225 PMCID: PMC10547463 DOI: 10.1093/infdis/jiad334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023] Open
Abstract
The National Center for Advancing Translational Sciences (NCATS) Assay Guidance Manual (AGM) Workshop on 3D Tissue Models for Antiviral Drug Development, held virtually on 7-8 June 2022, provided comprehensive coverage of critical concepts intended to help scientists establish robust, reproducible, and scalable 3D tissue models to study viruses with pandemic potential. This workshop was organized by NCATS, the National Institute of Allergy and Infectious Diseases, and the Bill and Melinda Gates Foundation. During the workshop, scientific experts from academia, industry, and government provided an overview of 3D tissue models' utility and limitations, use of existing 3D tissue models for antiviral drug development, practical advice, best practices, and case studies about the application of available 3D tissue models to infectious disease modeling. This report includes a summary of each workshop session as well as a discussion of perspectives and challenges related to the use of 3D tissues in antiviral drug discovery.
Collapse
Affiliation(s)
- Robert Jordan
- Bill and Melinda Gates Foundation, Seattle, Washington, USA
| | - Stephanie L Ford-Scheimer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Rodolfo M Alarcon
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | | | - Kyle R Brimacombe
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Sara Cherry
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Mindy I Davis
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Simon G P Funnell
- UK Health Security Agency, Salisbury, United Kingdom
- Quadram Institute Bioscience, Norwich, United Kingdom
| | - Lee Gehrke
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Abigail C Grossman
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Thomas Hartung
- Department of Environmental Health and Engineering, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Donald E Ingber
- Harvard Medical School, Boston, Massachusetts, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts, USA
- Harvard School of Engineering and Applied Sciences, Cambridge, Massachusetts, USA
- Boston Children's Hospital, Boston, Massachusetts, USA
| | - Nicole C Kleinstreuer
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle, North Carolina, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, California, USA
| | - Emily M Lee
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | | | - Thames E Pickett
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sasirekha Ramani
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | | | - Evi B Struble
- US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Zhengpeng Wan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Mark S Williams
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| | - Sarine Markossian
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, Maryland, USA
| |
Collapse
|
17
|
Luu RJ, Hoefler BC, Gard AL, Ritenour CR, Rogers MT, Kim ES, Coppeta JR, Cain BP, Isenberg BC, Azizgolshani H, Fajardo-Ramirez OR, García-Cardeña G, Lech MP, Tomlinson L, Charest JL, Williams C. Fibroblast activation in response to TGFβ1 is modulated by co-culture with endothelial cells in a vascular organ-on-chip platform. Front Mol Biosci 2023; 10:1160851. [PMID: 37577751 PMCID: PMC10421749 DOI: 10.3389/fmolb.2023.1160851] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/06/2023] [Indexed: 08/15/2023] Open
Abstract
Background: Tissue fibrosis is a major healthcare burden that affects various organs in the body for which no effective treatments exist. An underlying, emerging theme across organs and tissue types at early stages of fibrosis is the activation of pericytes and/or fibroblasts in the perivascular space. In hepatic tissue, it is well known that liver sinusoidal endothelial cells (EC) help maintain the quiescence of stellate cells, but whether this phenomenon holds true for other endothelial and perivascular cell types is not well studied. Methods: The goal of this work was to develop an organ-on-chip microvascular model to study the effect of EC co-culture on the activation of perivascular cells perturbed by the pro-fibrotic factor TGFβ1. A high-throughput microfluidic platform, PREDICT96, that was capable of imparting physiologically relevant fluid shear stress on the cultured endothelium was utilized. Results: We first studied the activation response of several perivascular cell types and selected a cell source, human dermal fibroblasts, that exhibited medium-level activation in response to TGFβ1. We also demonstrated that the PREDICT96 high flow pump triggered changes in select shear-responsive factors in human EC. We then found that the activation response of fibroblasts was significantly blunted in co-culture with EC compared to fibroblast mono-cultures. Subsequent studies with conditioned media demonstrated that EC-secreted factors play at least a partial role in suppressing the activation response. A Luminex panel and single cell RNA-sequencing study provided additional insight into potential EC-derived factors that could influence fibroblast activation. Conclusion: Overall, our findings showed that EC can reduce myofibroblast activation of perivascular cells in response to TGFβ1. Further exploration of EC-derived factors as potential therapeutic targets in fibrosis is warranted.
Collapse
Affiliation(s)
- Rebeccah J. Luu
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - B. Christopher Hoefler
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Ashley L. Gard
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | | | - Miles T. Rogers
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Ernest S. Kim
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Jonathan R. Coppeta
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Brian P. Cain
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Brett C. Isenberg
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Hesham Azizgolshani
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Oscar R. Fajardo-Ramirez
- Laboratory for Systems Mechanobiology, Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Guillermo García-Cardeña
- Laboratory for Systems Mechanobiology, Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | | | | | - Joseph L. Charest
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Corin Williams
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| |
Collapse
|
18
|
Chavarria D, Abbaspour A, Celestino N, Shah N, Sankar S, Baker AB. A high throughput blood-brain barrier model incorporating shear stress with improved predictive power for drug discovery. BIOMICROFLUIDICS 2023; 17:044105. [PMID: 37614679 PMCID: PMC10444201 DOI: 10.1063/5.0150887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/01/2023] [Indexed: 08/25/2023]
Abstract
The blood-brain barrier is a key structure regulating the health of the brain and access of drugs and pathogens to neural tissue. Shear stress is a key regulator of the blood-brain barrier; however, the commonly used multi-well vitro models of the blood-brain barrier do not incorporate shear stress. In this work, we designed and validated a high-throughput system for simulating the blood-brain barrier that incorporates physiological flow and incorporates an optimized cellular model of the blood-brain barrier. This system can perform assays of blood-brain barrier function with shear stress, with 48 independent assays simultaneously. Using the high throughput assay, we conducted drug screening assays to explore the effects of compounds for opening or closing blood-brain barrier. Our studies revealed that assays with shear stress were more predictive and were able to identify compounds known to modify the blood-brain barrier function while static assays were not. Overall, we demonstrate an optimized, high throughput assay for simulating the blood-brain barrier that incorporates shear stress and is practical for use in drug screening and other high throughput studies of toxicology.
Collapse
Affiliation(s)
- Daniel Chavarria
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | - Ali Abbaspour
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | - Natalie Celestino
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | - Nehali Shah
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, USA
| | | | - Aaron B. Baker
- Author to whom correspondence should be addressed:. Tel.:+512-232-7114
| |
Collapse
|
19
|
Marr EE, Mulhern TJ, Welch M, Keegan P, Caballero-Franco C, Johnson BG, Kasaian M, Azizgolshani H, Petrie T, Charest J, Wiellette E. A platform to reproducibly evaluate human colon permeability and damage. Sci Rep 2023; 13:8922. [PMID: 37264117 DOI: 10.1038/s41598-023-36020-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/27/2023] [Indexed: 06/03/2023] Open
Abstract
The intestinal epithelium comprises diverse cell types and executes many specialized functions as the primary interface between luminal contents and internal organs. A key function provided by the epithelium is maintenance of a barrier that protects the individual from pathogens, irritating luminal contents, and the microbiota. Disruption of this barrier can lead to inflammatory disease within the intestinal mucosa, and, in more severe cases, to sepsis. Animal models to study intestinal permeability are costly and not entirely predictive of human biology. Here we present a model of human colon barrier function that integrates primary human colon stem cells into Draper's PREDICT96 microfluidic organ-on-chip platform to yield a high-throughput system appropriate to predict damage and healing of the human colon epithelial barrier. We have demonstrated pharmacologically induced barrier damage measured by both a high throughput molecular permeability assay and transepithelial resistance. Using these assays, we developed an Inflammatory Bowel Disease-relevant model through cytokine induced damage that can support studies of disease mechanisms and putative therapeutics.
Collapse
Affiliation(s)
| | | | | | - Philip Keegan
- Draper, 555 Technology Sq., Cambridge, MA, 02139, USA
| | | | - Bryce G Johnson
- Pfizer Inflammation and Immunology, 1 Portland St., Cambridge, MA, 02139, USA
| | - Marion Kasaian
- Pfizer Inflammation and Immunology, 1 Portland St., Cambridge, MA, 02139, USA
| | | | | | | | | |
Collapse
|
20
|
Alvino VV, Mohammed KAK, Gu Y, Madeddu P. Approaches for the isolation and long-term expansion of pericytes from human and animal tissues. Front Cardiovasc Med 2023; 9:1095141. [PMID: 36704463 PMCID: PMC9873410 DOI: 10.3389/fcvm.2022.1095141] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/22/2022] [Indexed: 01/11/2023] Open
Abstract
Pericytes surround capillaries in every organ of the human body. They are also present around the vasa vasorum, the small blood vessels that supply the walls of larger arteries and veins. The clinical interest in pericytes is rapidly growing, with the recognition of their crucial roles in controlling vascular function and possible therapeutic applications in regenerative medicine. Nonetheless, discrepancies in methods used to define, isolate, and expand pericytes are common and may affect reproducibility. Separating pure pericyte preparations from the continuum of perivascular mesenchymal cells is challenging. Moreover, variations in functional behavior and antigenic phenotype in response to environmental stimuli make it difficult to formulate an unequivocal definition of bona fide pericytes. Very few attempts were made to develop pericytes as a clinical-grade product. Therefore, this review is devoted to appraising current methodologies' pros and cons and proposing standardization and harmonization improvements. We highlight the importance of developing upgraded protocols to create therapeutic pericyte products according to the regulatory guidelines for clinical manufacturing. Finally, we describe how integrating RNA-seq techniques with single-cell spatial analysis, and functional assays may help realize the full potential of pericytes in health, disease, and tissue repair.
Collapse
Affiliation(s)
| | - Khaled Abdelsattar Kassem Mohammed
- Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
- Department of Cardiothoracic Surgery, Faculty of Medicine, Assiut University, Asyut, Egypt
| | - Yue Gu
- Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | - Paolo Madeddu
- Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
21
|
Perfusion culture of endothelial cells under shear stress on microporous membrane in a pressure-driven microphysiological system. J Biosci Bioeng 2023; 135:79-85. [PMID: 36253250 DOI: 10.1016/j.jbiosc.2022.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/01/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022]
Abstract
This paper reports perfusion culture of human umbilical vein endothelial cells (HUVECs) on a microporous membrane in a pressure-driven microphysiological system (PD-MPS), which we developed previously as a multi-throughput perfusion culture platform. We designed fluidic culture unit with microporous membrane to culture HUVECs under fluidic shear stress and constructed a perfusion culture model in the PD-MPS platform. Four fluidic culture units were arranged in the microplate-sized device, which enables four-throughput assay for characterization of HUVECs under flow. Medium flow was generated above and below the membrane by sequential pneumatic pressure to apply physiological shear stress to HUVECs. HUVECs exhibited aligned morphology to the direction of the flow with shear stress of 11.5-17.7 dyn/cm2 under the flow condition, while they randomly aligned under static culture condition in a 6 well plate. We also observed 3.3- and 5.0-fold increase in the expression levels of the thrombomodulin and endothelial nitric oxide synthase mRNAs, respectively, under the flow condition in the PD-MPS compared to the static culture in 6 well plate. We also observed actin filament aligned to the direction of flow in HUVECs cultured under the flow condition.
Collapse
|
22
|
Yuan Y. Clinical Translation of Engineered Pulmonary Vascular Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1413:273-288. [PMID: 37195536 DOI: 10.1007/978-3-031-26625-6_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Diseases in pulmonary vasculature remain a major cause of morbidity and mortality worldwide. Numerous pre-clinical animal models were developed to understand lung vasculature during diseases and development. However, these systems are typically limited in their ability to represent human pathophysiology for the study of disease and drug mechanisms. In recent years, a growing number of studies have focused on developing in vitro experimental platforms that mimic human tissues/organs. In this chapter, we discuss the key components involved in developing engineered pulmonary vascular modeling systems and provide perspectives on ways to improve the translational potential of existing models.
Collapse
Affiliation(s)
- Yifan Yuan
- Department of Medicine (Pulmonary), Department of Anesthesiology, Yale University, New Haven, CT, USA.
| |
Collapse
|
23
|
Mansouri M, Ahmed A, Ahmad SD, McCloskey MC, Joshi IM, Gaborski TR, Waugh RE, McGrath JL, Day SW, Abhyankar VV. The Modular µSiM Reconfigured: Integration of Microfluidic Capabilities to Study In Vitro Barrier Tissue Models under Flow. Adv Healthc Mater 2022; 11:e2200802. [PMID: 35953453 PMCID: PMC9798530 DOI: 10.1002/adhm.202200802] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/01/2022] [Indexed: 01/28/2023]
Abstract
Microfluidic tissue barrier models have emerged to address the lack of physiological fluid flow in conventional "open-well" Transwell-like devices. However, microfluidic techniques have not achieved widespread usage in bioscience laboratories because they are not fully compatible with traditional experimental protocols. To advance barrier tissue research, there is a need for a platform that combines the key advantages of both conventional open-well and microfluidic systems. Here, a plug-and-play flow module is developed to introduce on-demand microfluidic flow capabilities to an open-well device that features a nanoporous membrane and live-cell imaging capabilities. The magnetic latching assembly of this design enables bi-directional reconfiguration and allows users to conduct an experiment in an open-well format with established protocols and then add or remove microfluidic capabilities as desired. This work also provides an experimentally-validated flow model to select flow conditions based on the experimental needs. As a proof-of-concept, flow-induced alignment of endothelial cells and the expression of shear-sensitive gene targets are demonstrated, and the different phases of neutrophil transmigration across a chemically stimulated endothelial monolayer under flow conditions are visualized. With these experimental capabilities, it is anticipated that both engineering and bioscience laboratories will adopt this reconfigurable design due to the compatibility with standard open-well protocols.
Collapse
Affiliation(s)
- Mehran Mansouri
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Adeel Ahmed
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - S. Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Indranil M. Joshi
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Thomas R. Gaborski
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Richard E. Waugh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Steven W. Day
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Vinay V. Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| |
Collapse
|
24
|
Kann SH, Shaughnessey EM, Coppeta JR, Azizgolshani H, Isenberg BC, Vedula EM, Zhang X, Charest JL. Measurement of oxygen consumption rates of human renal proximal tubule cells in an array of organ-on-chip devices to monitor drug-induced metabolic shifts. MICROSYSTEMS & NANOENGINEERING 2022; 8:109. [PMID: 36187891 PMCID: PMC9519964 DOI: 10.1038/s41378-022-00442-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 06/16/2023]
Abstract
Measurement of cell metabolism in moderate-throughput to high-throughput organ-on-chip (OOC) systems would expand the range of data collected for studying drug effects or disease in physiologically relevant tissue models. However, current measurement approaches rely on fluorescent imaging or colorimetric assays that are focused on endpoints, require labels or added substrates, and lack real-time data. Here, we integrated optical-based oxygen sensors in a high-throughput OOC platform and developed an approach for monitoring cell metabolic activity in an array of membrane bilayer devices. Each membrane bilayer device supported a culture of human renal proximal tubule epithelial cells on a porous membrane suspended between two microchannels and exposed to controlled, unidirectional perfusion and physiologically relevant shear stress for several days. For the first time, we measured changes in oxygen in a membrane bilayer format and used a finite element analysis model to estimate cell oxygen consumption rates (OCRs), allowing comparison with OCRs from other cell culture systems. Finally, we demonstrated label-free detection of metabolic shifts in human renal proximal tubule cells following exposure to FCCP, a drug known for increasing cell oxygen consumption, as well as oligomycin and antimycin A, drugs known for decreasing cell oxygen consumption. The capability to measure cell OCRs and detect metabolic shifts in an array of membrane bilayer devices contained within an industry standard microtiter plate format will be valuable for analyzing flow-responsive and physiologically complex tissues during drug development and disease research.
Collapse
Affiliation(s)
- Samuel H. Kann
- Draper Scholar, 555 Technology Square, Cambridge, MA 02139 USA
- Department of Mechanical Engineering, Boston University, 110 Cummington Mall, Boston, MA 02215 USA
| | - Erin M. Shaughnessey
- Draper Scholar, 555 Technology Square, Cambridge, MA 02139 USA
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155 USA
| | | | | | | | | | - Xin Zhang
- Department of Mechanical Engineering, Boston University, 110 Cummington Mall, Boston, MA 02215 USA
| | - Joseph L. Charest
- Draper, 555 Technology Square, Cambridge, MA 02139 USA
- Present Address: Biogen, 225 Binney Street, Cambridge, MA 02142 USA
| |
Collapse
|
25
|
Evaluation of rapid transepithelial electrical resistance (TEER) measurement as a metric of kidney toxicity in a high-throughput microfluidic culture system. Sci Rep 2022; 12:13182. [PMID: 35915212 PMCID: PMC9343646 DOI: 10.1038/s41598-022-16590-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/12/2022] [Indexed: 11/15/2022] Open
Abstract
Rapid non-invasive kidney-specific readouts are essential to maximizing the potential of microfluidic tissue culture platforms for drug-induced nephrotoxicity screening. Transepithelial electrical resistance (TEER) is a well-established technique, but it has yet to be evaluated as a metric of toxicity in a kidney proximal tubule (PT) model that recapitulates the high permeability of the native tissue and is also suitable for high-throughput screening. We utilized the PREDICT96 high-throughput microfluidic platform, which has rapid TEER measurement capability and multi-flow control, to evaluate the utility of TEER sensing for detecting cisplatin-induced toxicity in a human primary PT model under both mono- and co-culture conditions as well as two levels of fluid shear stress (FSS). Changes in TEER of PT-microvascular co-cultures followed a dose-dependent trend similar to that demonstrated by lactate dehydrogenase (LDH) cytotoxicity assays and were well-correlated with tight junction coverage after cisplatin exposure. Additionally, cisplatin-induced changes in TEER were detectable prior to increases in cell death in co-cultures. PT mono-cultures had a less differentiated phenotype and were not conducive to toxicity monitoring with TEER. The results of this study demonstrate that TEER has potential as a rapid, early, and label-free indicator of toxicity in microfluidic PT-microvascular co-culture models.
Collapse
|
26
|
Vaiana CA, Kim H, Cottet J, Oai K, Ge Z, Conforti K, King AM, Meyer AJ, Chen H, Voigt CA, Buie CR. Characterizing chemical signaling between engineered "microbial sentinels" in porous microplates. Mol Syst Biol 2022; 18:e10785. [PMID: 35315586 PMCID: PMC8938921 DOI: 10.15252/msb.202110785] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 01/23/2022] [Accepted: 01/25/2022] [Indexed: 11/11/2022] Open
Abstract
Living materials combine a material scaffold, that is often porous, with engineered cells that perform sensing, computing, and biosynthetic tasks. Designing such systems is difficult because little is known regarding signaling transport parameters in the material. Here, the development of a porous microplate is presented. Hydrogel barriers between wells have a porosity of 60% and a tortuosity factor of 1.6, allowing molecular diffusion between wells. The permeability of dyes, antibiotics, inducers, and quorum signals between wells were characterized. A "sentinel" strain was constructed by introducing orthogonal sensors into the genome of Escherichia coli MG1655 for IPTG, anhydrotetracycline, L-arabinose, and four quorum signals. The strain's response to inducer diffusion through the wells was quantified up to 14 mm, and quorum and antibacterial signaling were measured over 16 h. Signaling distance is dictated by hydrogel adsorption, quantified using a linear finite element model that yields adsorption coefficients from 0 to 0.1 mol m-3 . Parameters derived herein will aid the design of living materials for pathogen remediation, computation, and self-organizing biofilms.
Collapse
Affiliation(s)
- Christopher A Vaiana
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
- Synthetic Biology CenterDepartment of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Hyungseok Kim
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Jonathan Cottet
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Keiko Oai
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Zhifei Ge
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Kameron Conforti
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Andrew M King
- Synthetic Biology CenterDepartment of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Adam J Meyer
- Synthetic Biology CenterDepartment of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Haorong Chen
- Synthetic Biology CenterDepartment of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Christopher A Voigt
- Synthetic Biology CenterDepartment of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| | - Cullen R Buie
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMAUSA
| |
Collapse
|